Proteomics Reveals Ablation of Placental Growth Factor Inhibits the Insulin Resistance

Total Page:16

File Type:pdf, Size:1020Kb

Proteomics Reveals Ablation of Placental Growth Factor Inhibits the Insulin Resistance bioRxiv preprint doi: https://doi.org/10.1101/338368; this version posted June 4, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. 1 Proteomics reveals ablation of placental growth factor inhibits the insulin resistance 2 pathways in diabetic mouse retina 3 Madhu Sudhana Saddala1#, Anton Lennikov1# , Shibo Tang2,3, Hu Huang1,2,3* 4 5 1 Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, United States of America 6 2 Aier School of Ophthalmology, Central South University, Changsha, Hunan, China 7 3 Aier Eye Institute, Changsha, Hunan, China 8 9 *Corresponding author: 10 Hu Huang, PhD 11 Wilmer Eye Institute, Ophthalmology-Retinal Vascular Service 12 400 North Broadway St. Baltimore, MD, 21287 13 Hospital M017 Smith Building 14 Tel +1 (410)5020807 15 [email protected] 16 17 # Madhu Sudhana Saddala and Anton Lennikov have contributed equally to this work. 18 19 ORCID 20 Author 1: Madhu Sudhana Saddala ORCID: 0000-0002-6373-7080 21 Author 2: Anton Lennikov ORCID: 0000-0001-8625-1211 22 Author 3: Shibo Tang ORCID: 0000-0003-2737-6780 23 Author 4: Hu Huang ORCID: 0000-0003-2843-0320 24 1 bioRxiv preprint doi: https://doi.org/10.1101/338368; this version posted June 4, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. 25 Acknowledgments 26 The authors wish to acknowledge the contribution of: Lijuan Fan for technical assistance, Dmitry Rumyancev for 27 artwork design, and Jianjiang Hao for MS analysis. 28 Funding: 29 This work was supported by NIH grant (EY027824) 30 Authors Contributions: 31 The study was conceived and designed by M.S.S., A.L. and H.H. H.H. performed the animal handling, sample 32 collection and in vivo examinations. The manuscript was written by M.S.S, A.L.,H.H. and critically revised by H.H. 33 and S.T. All Authors reviewed and accepted the final version of the manuscript. 34 Additional Information 35 Financial interest statement: 36 The authors have no financial interests to disclose in relation to this paper. 37 38 Conflict of interest statement: 39 The authors have no conflict of interests to disclose in relation to this paper. 40 2 bioRxiv preprint doi: https://doi.org/10.1101/338368; this version posted June 4, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. 41 Abstract 42 The underlying molecular mechanisms that placental growth factor (PlGF) mediates the early complications at non- 43 proliferative diabetic retinopathy (DR) remain largely elusive. The objective of this study is to characterize 44 expression profile due to PlGF ablation in the retina of diabetic mice. The quantitative label-free proteomics was 45 carried out on retinal tissues collected from mouse strains (Akita; PlGF-/- and Akita.PlGF-/-). We have identified 46 3176 total proteins, and 107 were significantly different between the experimental groups, followed by gene 47 ontology, functional pathways, and protein-protein network interaction analysis. Gnb1, Gnb2, Gnb4, Gnai2, Gnao1, 48 Snap25, Stxbp1, Vamp2 and Gngt1 proteins are involved in insulin resistance pathways, which are down-regulated 49 in PlGF ablation in Akita diabetics (Akita.PlGF-/- vs. Akita), up-regulation in Akita vs. C57, PlGF-/- vs. C57. Prdx6, 50 Prdx5 (up-regulation) are known of antioxidant activity; Map2 is involved in neural protection pathways which are 51 up-regulated in Akita.PlGF-/- vs. Akita. Our results suggest that inhibition of insulin resistance pathway and the 52 enhancement of antioxidant defence and neural function may represent the potential mechanisms of anti-PlGF 53 compounds in the treatment of DR. 54 Total words: 175 55 56 57 58 59 Keywords: Diabetes, Akita mice, PlGF-knockout, Mass spectrometry, proteomics 60 3 bioRxiv preprint doi: https://doi.org/10.1101/338368; this version posted June 4, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. 61 1.0 Introduction 62 Diabetic retinopathy (DR), a sight-threatening microvascular complication of diabetes myelitis (DM), remains the 63 leading cause of vision loss worldwide in the adult population, especially in economically developed countries. (Lee 64 et al, 2015) With the increasing number of people with DM, the prevalence of DR and diabetic macular edema 65 (DME) is expected to grow. (Wild et al, 2004) Metabolic changes in the diabetic retina result in the altered 66 expression pattern of some mediators including growth factors, neurotrophic factors, cytokines/chemokines, 67 vasoactive agents, and inflammatory and adhesion molecules, resulting in vascular lesions and cell death. (Chen et 68 al, 2015; Kowluru & Mishra, 2015; Liu et al, 2015) Emerging evidence suggests that retinal neurodegeneration is an 69 early event in the pathogenesis of DR which could participate in the development of microvascular abnormalities. 70 (Barber, 2015; Simo et al, 2014) Placenta growth factor (PlGF), a member of VEGF family proteins, first discovered 71 in human placental cDNA in 1991. 72 In over two decades of scientific research and development have increased our understanding of the PlGF biological 73 function. Despite the high level of expression in placenta, the ablation of PlGF in mice did not compromise the 74 healthy embryonic development or adverse postnatal health effects. (Carmeliet & Jain, 2011) Delivery of 75 recombinant PlGF homodimer, PlGF-VEGFA heterodimer significantly promoted angiogenesis in ischemic 76 conditions through FLT1. (Luttun et al, 2002) Furthermore, many other cell types express PlGF in pathological 77 conditions, including retinal pigment epithelial cells (RPE). (Hollborn et al, 2006) This upregulation is due not 78 only to hypoxia but also from stimulus including nitric oxide (Mohammed et al, 2007), cytokines, as interleukin 79 1 (IL-1) and TNF-α (De Ceuninck et al, 2004), and transforming growth factor-β1 (TGF-β). (Yao et al, 2005) 80 The observation further confirmed the specific role of PlGF in pathological conditions that during pathological 81 angiogenesis endothelial cells over-express PlGF. (Ponticelli et al, 2008) 82 Recently our group has reported PlGF deletion in C57BL/6-Ins2Akita/J (Akita) mouse line, containing a dominant 83 mutation that induces spontaneous diabetes with a rapid onset. (Barber et al, 2005) Ablation of PlGF in the diabetic 84 mice resulted in an decreased expression of diabetes-activated hypoxia-inducible factor (HIF)1α, vascular 85 endothelial growth factor (VEGF) pathway, including expression of HIF1α, VEGF, VEGFR1–3, and the extent of 86 phospho (p)-VEGFR1, p-VEGFR2, and p–endothelial nitric oxide synthase, in the retinas of diabetic PlGF−/− mice. 87 Without a noticeable effect on glucose balance or expression of intercellular adhesion molecule-1, vascular cell 88 adhesion molecule-1, CD11b, and CD18 (Huang et al, 2015a). 89 While many functions and biological roles of PlGF are still currently unknown, the transition to human patients with 90 two phase II clinical trials of anti-PlGF recombinant monoclonal antibody in human DR patients is currently 91 underway. (NCT03071068; NCT03499223; ThromboGenics, 2018) Use of PlGF antibodies in humans presents a 4 bioRxiv preprint doi: https://doi.org/10.1101/338368; this version posted June 4, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. 92 challenge of better understanding the functions and pathways involved in PlGF knock out on the proteome scale. 93 Label-free mass spectrometry (LFMS) is a widely used tool for protein identification and quantification it is a gel- 94 free method allowing to conduct whole proteome analysis without the use of isotopic labeling (Luber et al, 2010). 95 Furthermore, as DR affects the expression of many commonly used “housekeeping” proteins such as ACTB and 96 Tubulin, MS approach resolve this issue as proteins are identified by the number of peptide sequences rather than 97 any relative quantification that requires a “housekeeping” protein (Rocha-Martins et al, 2012), (Li & Shen, 2013). In 98 the current study, we used label-free quantitative proteomics analysis to study retinal protein extracts from three 99 genetically modified mouse strains with diabetic and PlGF knockout condition as well as a combination of both to 100 further elucidate the molecular mechanisms of PlGF knockout playing beneficial role in DR on the proteome wide 101 level. We have identified 3176 total proteins, and 107 were significantly different between the experimental groups 102 (p<0.05). 103 104 1.0 Results 105 2.1 Animals and diabetic conditions. 106 Four 5-6 months old female mice were selected from each strain: C57BL/6-Ins2<Akita>/J.PlGF-/- (Akita.PlGF-/-), 107 PlGF-/-, C57BL/6-Ins2<Akita>/J (Akita), and C57BL/6J (C57) for this study. Graphical abstract of the animal 108 breeding program is presented in Figure 1A. Animals blood glucose, levels of glycated hemoglobin (HbA1c) and 109 body weight are presented in Table 1. All Akita mice have demonstrated significant increase of blood glucose (BG) 110 levels (p<0.001), HbA1c (p<0.05) and a decrease in body weight (p<0.01) when compared with C57 control animals 111 of the same age. Lack of PlGF did not affect blood glucose (p>0.05) levels of glycated hemoglobin (HbA1c) 112 (p>0.05) and body weight (p>0.05) for Akita.PlGF-/- vs. Akita. There was no significant difference in any of the 113 parameters in PlGF-/- vs. C57 mice of the same age. Retinal protein extracts 4 samples per group were trypsin 114 digested and subjected to the LC/MS/MS analysis (Figure 1C,D).
Recommended publications
  • Whole-Genome Microarray Detects Deletions and Loss of Heterozygosity of Chromosome 3 Occurring Exclusively in Metastasizing Uveal Melanoma
    Anatomy and Pathology Whole-Genome Microarray Detects Deletions and Loss of Heterozygosity of Chromosome 3 Occurring Exclusively in Metastasizing Uveal Melanoma Sarah L. Lake,1 Sarah E. Coupland,1 Azzam F. G. Taktak,2 and Bertil E. Damato3 PURPOSE. To detect deletions and loss of heterozygosity of disease is fatal in 92% of patients within 2 years of diagnosis. chromosome 3 in a rare subset of fatal, disomy 3 uveal mela- Clinical and histopathologic risk factors for UM metastasis noma (UM), undetectable by fluorescence in situ hybridization include large basal tumor diameter (LBD), ciliary body involve- (FISH). ment, epithelioid cytomorphology, extracellular matrix peri- ϩ ETHODS odic acid-Schiff-positive (PAS ) loops, and high mitotic M . Multiplex ligation-dependent probe amplification 3,4 5 (MLPA) with the P027 UM assay was performed on formalin- count. Prescher et al. showed that a nonrandom genetic fixed, paraffin-embedded (FFPE) whole tumor sections from 19 change, monosomy 3, correlates strongly with metastatic death, and the correlation has since been confirmed by several disomy 3 metastasizing UMs. Whole-genome microarray analy- 3,6–10 ses using a single-nucleotide polymorphism microarray (aSNP) groups. Consequently, fluorescence in situ hybridization were performed on frozen tissue samples from four fatal dis- (FISH) detection of chromosome 3 using a centromeric probe omy 3 metastasizing UMs and three disomy 3 tumors with Ͼ5 became routine practice for UM prognostication; however, 5% years’ metastasis-free survival. to 20% of disomy 3 UM patients unexpectedly develop metas- tases.11 Attempts have therefore been made to identify the RESULTS. Two metastasizing UMs that had been classified as minimal region(s) of deletion on chromosome 3.12–15 Despite disomy 3 by FISH analysis of a small tumor sample were found these studies, little progress has been made in defining the key on MLPA analysis to show monosomy 3.
    [Show full text]
  • Seq2pathway Vignette
    seq2pathway Vignette Bin Wang, Xinan Holly Yang, Arjun Kinstlick May 19, 2021 Contents 1 Abstract 1 2 Package Installation 2 3 runseq2pathway 2 4 Two main functions 3 4.1 seq2gene . .3 4.1.1 seq2gene flowchart . .3 4.1.2 runseq2gene inputs/parameters . .5 4.1.3 runseq2gene outputs . .8 4.2 gene2pathway . 10 4.2.1 gene2pathway flowchart . 11 4.2.2 gene2pathway test inputs/parameters . 11 4.2.3 gene2pathway test outputs . 12 5 Examples 13 5.1 ChIP-seq data analysis . 13 5.1.1 Map ChIP-seq enriched peaks to genes using runseq2gene .................... 13 5.1.2 Discover enriched GO terms using gene2pathway_test with gene scores . 15 5.1.3 Discover enriched GO terms using Fisher's Exact test without gene scores . 17 5.1.4 Add description for genes . 20 5.2 RNA-seq data analysis . 20 6 R environment session 23 1 Abstract Seq2pathway is a novel computational tool to analyze functional gene-sets (including signaling pathways) using variable next-generation sequencing data[1]. Integral to this tool are the \seq2gene" and \gene2pathway" components in series that infer a quantitative pathway-level profile for each sample. The seq2gene function assigns phenotype-associated significance of genomic regions to gene-level scores, where the significance could be p-values of SNPs or point mutations, protein-binding affinity, or transcriptional expression level. The seq2gene function has the feasibility to assign non-exon regions to a range of neighboring genes besides the nearest one, thus facilitating the study of functional non-coding elements[2]. Then the gene2pathway summarizes gene-level measurements to pathway-level scores, comparing the quantity of significance for gene members within a pathway with those outside a pathway.
    [Show full text]
  • Supplemental Materials Figure 1. 80 Genes Most Highly Differentially Expressed Comparing Dedifferentiated to Well- Differentiated Liposarcoma
    Supplemental Materials Figure 1. 80 genes most highly differentially expressed comparing dedifferentiated to well- differentiated liposarcoma Figure 2. Validation of microa CDC2, RACGAP1, FGFR-2, MAD2 and CITED1 200 CDK4 by Liposarcoma Subtype 16 0 12 0 80 Expression40 Level 0 Nl Fat rray data by quantitative RT-P Well Diff 16 0 p16 by Liposarcoma Subtype Dediff 12 0 80 Myxoid Expression40 Level Round Cell 0 12 0 MDM2 by Liposarcoma Subtype 10 0 Pleomorphic Nl Fat 80 60 40 Expression Level 20 RACGAP1 by LiposarcomaWell Diff Subtype 0 70 CR for genes CDK4, MDM2, p16, 60 Dediff 50 Nl Fat 40 30 Myxoid Expression20 Level Well Diff 10 50 0 Round Cell CDC2 by Liposarcoma Subtype Dediff 40 Nl Fat Pleomorphic 30 Myxoid 20 Expression Level Well Diff 10 MAD2L1 by Liposarcoma Subtype Round Cell 60 0 50 Dediff Pleomorphic 40 Nl Fat 30 Myxoid 20 Expression Level Well Diff 10 FGFR-2 by Liposarcoma Subtype 0 Round Cell 200 16 0 Dediff Nl Fat Pleomorphic 12 0 Myxoid 80 Expression Level Well Diff 40 Round Cell 0 Dediff Pleomorphic Nl Fat Myxoid Well Diff CITED1 by Liposarcoma Subtype Round Cell 10 0 80 Dediff Pleomorphic 60 40 Myxoid Expression Level 20 0 Round Cell Nl Fat Pleomorphic Well Diff Dediff Myxoid Round Cell Pleomorphic Table 1. 142 Gene classifier for liposarcoma subtype The genes used for each pair wise subtype comparison are grouped together. The flag column indicates which genes are unique to each subtype comparison. The values show the mean expression levels (actually the mean of the log expression levels was computed and than transformed back to absolute expression level).
    [Show full text]
  • Stelios Pavlidis3, Matthew Loza3, Fred Baribaud3, Anthony
    Supplementary Data Th2 and non-Th2 molecular phenotypes of asthma using sputum transcriptomics in UBIOPRED Chih-Hsi Scott Kuo1.2, Stelios Pavlidis3, Matthew Loza3, Fred Baribaud3, Anthony Rowe3, Iaonnis Pandis2, Ana Sousa4, Julie Corfield5, Ratko Djukanovic6, Rene 7 7 8 2 1† Lutter , Peter J. Sterk , Charles Auffray , Yike Guo , Ian M. Adcock & Kian Fan 1†* # Chung on behalf of the U-BIOPRED consortium project team 1Airways Disease, National Heart & Lung Institute, Imperial College London, & Biomedical Research Unit, Biomedical Research Unit, Royal Brompton & Harefield NHS Trust, London, United Kingdom; 2Department of Computing & Data Science Institute, Imperial College London, United Kingdom; 3Janssen Research and Development, High Wycombe, Buckinghamshire, United Kingdom; 4Respiratory Therapeutic Unit, GSK, Stockley Park, United Kingdom; 5AstraZeneca R&D Molndal, Sweden and Areteva R&D, Nottingham, United Kingdom; 6Faculty of Medicine, Southampton University, Southampton, United Kingdom; 7Faculty of Medicine, University of Amsterdam, Amsterdam, Netherlands; 8European Institute for Systems Biology and Medicine, CNRS-ENS-UCBL, Université de Lyon, France. †Contributed equally #Consortium project team members are listed under Supplementary 1 Materials *To whom correspondence should be addressed: [email protected] 2 List of the U-BIOPRED Consortium project team members Uruj Hoda & Christos Rossios, Airways Disease, National Heart & Lung Institute, Imperial College London, UK & Biomedical Research Unit, Biomedical Research Unit, Royal
    [Show full text]
  • Investigating Cone Photoreceptor Development Using Patient-Derived NRL Null Retinal Organoids
    ARTICLE https://doi.org/10.1038/s42003-020-0808-5 OPEN Investigating cone photoreceptor development using patient-derived NRL null retinal organoids Alyssa Kallman1,11, Elizabeth E. Capowski 2,11, Jie Wang 3, Aniruddha M. Kaushik4, Alex D. Jansen2, Kimberly L. Edwards2, Liben Chen4, Cynthia A. Berlinicke3, M. Joseph Phillips2,5, Eric A. Pierce6, Jiang Qian3, ✉ ✉ Tza-Huei Wang4,7, David M. Gamm2,5,8 & Donald J. Zack 1,3,9,10 1234567890():,; Photoreceptor loss is a leading cause of blindness, but mechanisms underlying photoreceptor degeneration are not well understood. Treatment strategies would benefit from improved understanding of gene-expression patterns directing photoreceptor development, as many genes are implicated in both development and degeneration. Neural retina leucine zipper (NRL) is critical for rod photoreceptor genesis and degeneration, with NRL mutations known to cause enhanced S-cone syndrome and retinitis pigmentosa. While murine Nrl loss has been characterized, studies of human NRL can identify important insights for human retinal development and disease. We utilized iPSC organoid models of retinal development to molecularly define developmental alterations in a human model of NRL loss. Consistent with the function of NRL in rod fate specification, human retinal organoids lacking NRL develop S- opsin dominant photoreceptor populations. We report generation of two distinct S-opsin expressing populations in NRL null retinal organoids and identify MEF2C as a candidate regulator of cone development. 1 Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, USA. 2 Waisman Center, University of Wisconsin-Madison, Madison, USA. 3 Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, USA.
    [Show full text]
  • A Computational Approach for Defining a Signature of Β-Cell Golgi Stress in Diabetes Mellitus
    Page 1 of 781 Diabetes A Computational Approach for Defining a Signature of β-Cell Golgi Stress in Diabetes Mellitus Robert N. Bone1,6,7, Olufunmilola Oyebamiji2, Sayali Talware2, Sharmila Selvaraj2, Preethi Krishnan3,6, Farooq Syed1,6,7, Huanmei Wu2, Carmella Evans-Molina 1,3,4,5,6,7,8* Departments of 1Pediatrics, 3Medicine, 4Anatomy, Cell Biology & Physiology, 5Biochemistry & Molecular Biology, the 6Center for Diabetes & Metabolic Diseases, and the 7Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202; 2Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202; 8Roudebush VA Medical Center, Indianapolis, IN 46202. *Corresponding Author(s): Carmella Evans-Molina, MD, PhD ([email protected]) Indiana University School of Medicine, 635 Barnhill Drive, MS 2031A, Indianapolis, IN 46202, Telephone: (317) 274-4145, Fax (317) 274-4107 Running Title: Golgi Stress Response in Diabetes Word Count: 4358 Number of Figures: 6 Keywords: Golgi apparatus stress, Islets, β cell, Type 1 diabetes, Type 2 diabetes 1 Diabetes Publish Ahead of Print, published online August 20, 2020 Diabetes Page 2 of 781 ABSTRACT The Golgi apparatus (GA) is an important site of insulin processing and granule maturation, but whether GA organelle dysfunction and GA stress are present in the diabetic β-cell has not been tested. We utilized an informatics-based approach to develop a transcriptional signature of β-cell GA stress using existing RNA sequencing and microarray datasets generated using human islets from donors with diabetes and islets where type 1(T1D) and type 2 diabetes (T2D) had been modeled ex vivo. To narrow our results to GA-specific genes, we applied a filter set of 1,030 genes accepted as GA associated.
    [Show full text]
  • Predicting Coupling Probabilities of G-Protein Coupled Receptors Gurdeep Singh1,2,†, Asuka Inoue3,*,†, J
    Published online 30 May 2019 Nucleic Acids Research, 2019, Vol. 47, Web Server issue W395–W401 doi: 10.1093/nar/gkz392 PRECOG: PREdicting COupling probabilities of G-protein coupled receptors Gurdeep Singh1,2,†, Asuka Inoue3,*,†, J. Silvio Gutkind4, Robert B. Russell1,2,* and Francesco Raimondi1,2,* 1CellNetworks, Bioquant, Heidelberg University, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany, 2Biochemie Zentrum Heidelberg (BZH), Heidelberg University, Im Neuenheimer Feld 328, 69120 Heidelberg, Germany, 3Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan and 4Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA Received February 10, 2019; Revised April 13, 2019; Editorial Decision April 24, 2019; Accepted May 01, 2019 ABSTRACT great use in tinkering with signalling pathways in living sys- tems (5). G-protein coupled receptors (GPCRs) control multi- Ligand binding to GPCRs induces conformational ple physiological states by transducing a multitude changes that lead to binding and activation of G-proteins of extracellular stimuli into the cell via coupling to situated on the inner cell membrane. Most of mammalian intra-cellular heterotrimeric G-proteins. Deciphering GPCRs couple with more than one G-protein giving each which G-proteins couple to each of the hundreds receptor a distinct coupling profile (6) and thus specific of GPCRs present in a typical eukaryotic organism downstream cellular responses. Determining these coupling is therefore critical to understand signalling. Here, profiles is critical to understand GPCR biology and phar- we present PRECOG (precog.russelllab.org): a web- macology. Despite decades of research and hundreds of ob- server for predicting GPCR coupling, which allows served interactions, coupling information is still missing for users to: (i) predict coupling probabilities for GPCRs many receptors and sequence determinants of coupling- specificity are still largely unknown.
    [Show full text]
  • Cellular and Molecular Signatures in the Disease Tissue of Early
    Cellular and Molecular Signatures in the Disease Tissue of Early Rheumatoid Arthritis Stratify Clinical Response to csDMARD-Therapy and Predict Radiographic Progression Frances Humby1,* Myles Lewis1,* Nandhini Ramamoorthi2, Jason Hackney3, Michael Barnes1, Michele Bombardieri1, Francesca Setiadi2, Stephen Kelly1, Fabiola Bene1, Maria di Cicco1, Sudeh Riahi1, Vidalba Rocher-Ros1, Nora Ng1, Ilias Lazorou1, Rebecca E. Hands1, Desiree van der Heijde4, Robert Landewé5, Annette van der Helm-van Mil4, Alberto Cauli6, Iain B. McInnes7, Christopher D. Buckley8, Ernest Choy9, Peter Taylor10, Michael J. Townsend2 & Costantino Pitzalis1 1Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK. Departments of 2Biomarker Discovery OMNI, 3Bioinformatics and Computational Biology, Genentech Research and Early Development, South San Francisco, California 94080 USA 4Department of Rheumatology, Leiden University Medical Center, The Netherlands 5Department of Clinical Immunology & Rheumatology, Amsterdam Rheumatology & Immunology Center, Amsterdam, The Netherlands 6Rheumatology Unit, Department of Medical Sciences, Policlinico of the University of Cagliari, Cagliari, Italy 7Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G12 8TA, UK 8Rheumatology Research Group, Institute of Inflammation and Ageing (IIA), University of Birmingham, Birmingham B15 2WB, UK 9Institute of
    [Show full text]
  • Macropinocytosis Requires Gal-3 in a Subset of Patient-Derived Glioblastoma Stem Cells
    ARTICLE https://doi.org/10.1038/s42003-021-02258-z OPEN Macropinocytosis requires Gal-3 in a subset of patient-derived glioblastoma stem cells Laetitia Seguin1,8, Soline Odouard2,8, Francesca Corlazzoli 2,8, Sarah Al Haddad2, Laurine Moindrot2, Marta Calvo Tardón3, Mayra Yebra4, Alexey Koval5, Eliana Marinari2, Viviane Bes3, Alexandre Guérin 6, Mathilde Allard2, Sten Ilmjärv6, Vladimir L. Katanaev 5, Paul R. Walker3, Karl-Heinz Krause6, Valérie Dutoit2, ✉ Jann N. Sarkaria 7, Pierre-Yves Dietrich2 & Érika Cosset 2 Recently, we involved the carbohydrate-binding protein Galectin-3 (Gal-3) as a druggable target for KRAS-mutant-addicted lung and pancreatic cancers. Here, using glioblastoma patient-derived stem cells (GSCs), we identify and characterize a subset of Gal-3high glio- 1234567890():,; blastoma (GBM) tumors mainly within the mesenchymal subtype that are addicted to Gal-3- mediated macropinocytosis. Using both genetic and pharmacologic inhibition of Gal-3, we showed a significant decrease of GSC macropinocytosis activity, cell survival and invasion, in vitro and in vivo. Mechanistically, we demonstrate that Gal-3 binds to RAB10, a member of the RAS superfamily of small GTPases, and β1 integrin, which are both required for macro- pinocytosis activity and cell survival. Finally, by defining a Gal-3/macropinocytosis molecular signature, we could predict sensitivity to this dependency pathway and provide proof-of- principle for innovative therapeutic strategies to exploit this Achilles’ heel for a significant and unique subset of GBM patients. 1 University Côte d’Azur, CNRS UMR7284, INSERM U1081, Institute for Research on Cancer and Aging (IRCAN), Nice, France. 2 Laboratory of Tumor Immunology, Department of Oncology, Center for Translational Research in Onco-Hematology, Swiss Cancer Center Léman (SCCL), Geneva University Hospitals, University of Geneva, Geneva, Switzerland.
    [Show full text]
  • CLIP-Seq and Massively Parallel Functional Analysis of the CELF6
    bioRxiv preprint doi: https://doi.org/10.1101/401604; this version posted August 27, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. CLIP-Seq and massively parallel functional analysis of the CELF6 RNA binding protein reveals a role in destabilizing synaptic gene mRNAs through interaction with 3’UTR elements in vivo Michael A. Rieger1,2, Dana M. King1, Barak A. Cohen1, Joseph D. Dougherty1,2 1Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA 2Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA Contact: Dr. Joseph D. Dougherty Washington University School of Medicine Department of Genetics Campus Box 8232 4566 Scott Ave. St. Louis, Mo. 63110-1093 (314) 286-0752 [email protected] bioRxiv preprint doi: https://doi.org/10.1101/401604; this version posted August 27, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Abstract CELF6 is an RNA-binding protein in a family of proteins with roles in human health and disease, however little is known about the mRNA targets or in vivo function of this protein. We utilized HITS-CLIP/CLIP-Seq to identify, for the first time, in vivo targets of CELF6 and identify hundreds of transcripts bound by CELF6 in the brain. We found these are disproportionately mRNAs coding for synaptic proteins. We then conducted extensive functional validation of these targets, testing greater than 400 CELF6 bound sequence elements for their activity, applying a massively parallel reporter assay framework to evaluation of the CLIP data.
    [Show full text]
  • Haplotypes Are Associated with Susceptibility To
    www.nature.com/scientificreports OPEN Serpin peptidase inhibitor (SERPINB5) haplotypes are associated with susceptibility to Received: 02 March 2016 Accepted: 05 May 2016 hepatocellular carcinoma Published: 25 May 2016 Shun-Fa Yang1,2, Chao-Bin Yeh3,4, Ying-Erh Chou2,5, Hsiang-Lin Lee1,6 & Yu-Fan Liu7,8 Hepatocellular carcinoma (HCC) represents the second leading cause of cancer-related death worldwide. The serpin peptidase inhibitor SERPINB5 is a tumour-suppressor gene that promotes the development of various cancers in humans. However, whether SERPINB5 gene variants play a role in HCC susceptibility remains unknown. In this study, we genotyped 6 SNPs of the SERPINB5 gene in an independent cohort from a replicate population comprising 302 cases and 590 controls. Additionally, patients who had at least one rs2289520 C allele in SERPINB5 tended to exhibit better liver function than patients with genotype GG (Child-Pugh grade A vs. B or C; P = 0.047). Next, haplotype blocks were reconstructed according to the linkage disequilibrium structure of the SERPINB5 gene. A haplotype “C-C-C” (rs17071138 + rs3744941 + rs8089204) in SERPINB5-correlated promoter showed a significant association with an increased HCC risk (AOR = 1.450; P = 0.031). Haplotypes “T-C-A” and “C-C-C” (rs2289519 + rs2289520 + rs1455555) located in the SERPINB5 coding region had a decreased (AOR = 0.744; P = 0.031) and increased (AOR = 1.981; P = 0.001) HCC risk, respectively. Finally, an additional integrated in silico analysis confirmed that these SNPs affectedSERPINB5 expression and protein stability, which significantly correlated with tumour expression and subsequently with tumour development and aggressiveness.
    [Show full text]
  • Supplementary Table 1. Pain and PTSS Associated Genes (N = 604
    Supplementary Table 1. Pain and PTSS associated genes (n = 604) compiled from three established pain gene databases (PainNetworks,[61] Algynomics,[52] and PainGenes[42]) and one PTSS gene database (PTSDgene[88]). These genes were used in in silico analyses aimed at identifying miRNA that are predicted to preferentially target this list genes vs. a random set of genes (of the same length). ABCC4 ACE2 ACHE ACPP ACSL1 ADAM11 ADAMTS5 ADCY5 ADCYAP1 ADCYAP1R1 ADM ADORA2A ADORA2B ADRA1A ADRA1B ADRA1D ADRA2A ADRA2C ADRB1 ADRB2 ADRB3 ADRBK1 ADRBK2 AGTR2 ALOX12 ANO1 ANO3 APOE APP AQP1 AQP4 ARL5B ARRB1 ARRB2 ASIC1 ASIC2 ATF1 ATF3 ATF6B ATP1A1 ATP1B3 ATP2B1 ATP6V1A ATP6V1B2 ATP6V1G2 AVPR1A AVPR2 BACE1 BAMBI BDKRB2 BDNF BHLHE22 BTG2 CA8 CACNA1A CACNA1B CACNA1C CACNA1E CACNA1G CACNA1H CACNA2D1 CACNA2D2 CACNA2D3 CACNB3 CACNG2 CALB1 CALCRL CALM2 CAMK2A CAMK2B CAMK4 CAT CCK CCKAR CCKBR CCL2 CCL3 CCL4 CCR1 CCR7 CD274 CD38 CD4 CD40 CDH11 CDK5 CDK5R1 CDKN1A CHRM1 CHRM2 CHRM3 CHRM5 CHRNA5 CHRNA7 CHRNB2 CHRNB4 CHUK CLCN6 CLOCK CNGA3 CNR1 COL11A2 COL9A1 COMT COQ10A CPN1 CPS1 CREB1 CRH CRHBP CRHR1 CRHR2 CRIP2 CRYAA CSF2 CSF2RB CSK CSMD1 CSNK1A1 CSNK1E CTSB CTSS CX3CL1 CXCL5 CXCR3 CXCR4 CYBB CYP19A1 CYP2D6 CYP3A4 DAB1 DAO DBH DBI DICER1 DISC1 DLG2 DLG4 DPCR1 DPP4 DRD1 DRD2 DRD3 DRD4 DRGX DTNBP1 DUSP6 ECE2 EDN1 EDNRA EDNRB EFNB1 EFNB2 EGF EGFR EGR1 EGR3 ENPP2 EPB41L2 EPHB1 EPHB2 EPHB3 EPHB4 EPHB6 EPHX2 ERBB2 ERBB4 EREG ESR1 ESR2 ETV1 EZR F2R F2RL1 F2RL2 FAAH FAM19A4 FGF2 FKBP5 FLOT1 FMR1 FOS FOSB FOSL2 FOXN1 FRMPD4 FSTL1 FYN GABARAPL1 GABBR1 GABBR2 GABRA2 GABRA4
    [Show full text]